Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

Combined treatment with lexatumumab and irradiation leads to strongly increased long term tumour control under normoxic and hypoxic conditions

Combined treatment with lexatumumab and irradiation leads to strongly increased long term tumour... Purpose: The combination of ionizing radiation with the pro-apoptotic TRAIL receptor antibody lexatumumab has been shown to exert considerable synergistic apoptotic effects in vitro and in short term growth delay assays. To clarify the relevance of these effects on local tumour control long-term experiments using a colorectal xenograft model were conducted. Materials and methods: Colo205-xenograft bearing NMRI (nu/nu) nude mice were treated with fractionated irradiation (5× 3 Gy, d1-5) and lexatumumab (0.75 mg/kg, d1, 4 and 8). The tumour bearing hind limbs were irradiated with graded single top up doses at d8 under normoxic (ambient) and acute hypoxic (clamped) conditions. Experimental animals were observed for 270 days. Growth delay and local tumour control were end points of the study. Statistical analysis of the experiments included evaluation of tumour regrowth and local tumour control. Results: Combined treatment with irradiation and lexatumumab led to a pronounced tumour regrowth-delay when compared to irradiation alone. The here presented long-term experiments revealed a highly significant rise of local tumour control for normoxic (ambient) (p = 0. 000006) and hypoxic treatment (p = 0. 000030). Conclusion: Our data show that a combination of the pro-apoptotic antibody lexatumumab with irradiation reduces tumour regrowth and leads to a highly increased local tumour control in a nude mouse model. This substantial effect was observed under ambient and more pronounced under hypoxic conditions. related apoptosis inducing ligand) receptor 2 (TRAIL-R2) Background Lexatumumab is a fully human agonistic antibody with a induced apoptosis. Although TRAIL-R2 stimulation alone distinct tumour cell specifity via activation of TRAIL (TNF- is highly effective in a wide range of cancer cell lines, effi- Page 1 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 cacy can be increased by combination with other gyro- Materials and methods static drugs (for review see [1]). We have already shown Animals and tumours Immunodeficient NMRI-(nu/nu)-nude mice were pur- that a combined treatment with TRAIL and irradiation exerts highly synergistic effects regarding apoptosis induc- chased from a specific pathogen free colony at the Univer- tion. This enhanced efficacy was detectable in various sity of Essen (Germany) at the age of 4-6 weeks. Animals solid tumour cell lines and lymphoid tumour cells[2,3]. were kept in an individually ventilated cage rack system (Techniplast, Italy) and fed with sterile high calorie labo- Since discovery of TRAIL and its receptors in 1997 a panel ratory food (Sniff, Germany). Drank water was supple- of agonistic antibodies for TRAIL-receptors R1 and R2 mented by chlorotetracycline and potassium sorbate have been developed and tested in clinical phase I and II acidified to a pH of 3.0 with hydrochloric acid. trials [4-18]. However, up to now only little data are avail- able concerning interaction of agonistic TRAIL receptor The Colo205 tumour cell line (established from a colorec- antibodies and irradiation ([7,19,20]. Besides our recently tal adenocarcinoma) was acquired from ATCC (Bethesda, published report no data on experiments with a combina- MD, USA). In NMRI-(nu/nu)-nude mice Colo205 cells tion of a fully human TRAIL receptor antibody and irradi- form solid, roundly shaped tumours without indication ation have been published[21]. for metastasis. Combining mapatumumab or lexatumumab with irradi- Transplantation and experimental design ation, we have demonstrated that this combination exerts Tumour lumps of about 2 mm diameter from a source strong additive and synergistic effects on apoptosis induc- tumour were implanted subcutaneously into the right tion in vitro and in short-term growth delay experi- hind limb of 6-10 week old animals. Approximately 2-3 ments[10]. However, to proof that induction of apoptosis weeks after transplantation tumour growth was measura- evidently translates into definitive tumour stem cell erad- ble. Tumour size was quantified with calipers in two per- ication long-term experiments with local tumour control pendicular diameters. The tumour volume (V) was as primary endpoint might provide a reliable model for calculated as V = (a × b )/2, where a and b are the long axis clinical potency [22-26]. and the short axis, respectively. Scoring of tumour sizes took place three times per week before start of treatment. Therefore, we decided to perform long-term experiments Body weight was monitored once a week. in a nude mouse xenograft model. As radiation sensitivity becomes affected by limiting intratumoural hypoxia we The median tumour volume at the start of experiments run experiments under both ambient and hypoxic condi- was 116 ± 31 mm . Animals were randomly allocated to tions to mimic realistic tumour conditions[27]. 24 treatment arms (scheme see Figure 1): lexatumumab at day 1, 4 and 8 (0.75 mg/kg body weight intraperitoneally Taken together, our experimental series was designed to (i.p.)) alone, fractioned radiotherapy (5 × 3 Gy within five confirm the striking principle that radiation mediated subsequent days) alone. Single dose top up irradiations TRAIL sensitization effectively increases long-term local (0, 10.0, 14.5, 21.0, 30.4, 44.2 Gy) were performed on day tumour control. 8. Combined treatment was performed at day 1, 4 and 8 with lexatumumab (0.75 mg/kg) (figure 1). Control ani- graded top up dose (0-44,2 Gy) lexatumumab (0.75 mg/kg)/ θθ ±±±± irradiation d1 d4 d5 d8 d270 E Figure 1 xperimental design Experimental design. Small bolt = fractionated irradiation at d 1-5, large bolt = graded top up doses 0-44.2 Gy (under ambi- ent/hypoxic conditions, depending on stratification), small arrowhead: application of lexatumumab (0.75 mg/kg body weight), d = day. Page 2 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 mals were treated only with an i.p. injection of medium Figure 2 shows a chronological sequence of the impressive without antibody or irradiation. tumour regression after treatment with lexatumumab (0.75 mg/kg) for one test animal, exemplarily. Obviously, To minimize toxic side effects and to apply high irradia- tumour growth reduction started after the second applica- tion doses in an easy comparable, time saving schedule we tion i.p., already. However, lacking consolidating irradia- choose a combination of fractionated and graded single tion in this example tumour regrowth is evident four high dose (top up) irradiation. 3 Gy single dose was cho- weeks after start of treatment. sen for fractionated irradiation based on previous experi- ments (Marini et al., Oncogene 2006). Fractionated However, combination of very low doses of irradiation irradiation of tumours was applied in inhalation (Isoflu- with lexatumumab led to an unexpected high local rane) narcosis. Top up irradiation under ambient condi- tumour rate, already. Tumour regrowth after combined tions or under clamped hypoxia was performed with i.p. treatment was observed in less than 50% of the animals. narcosis (fentanyl, midazolam, medetomidine), as rec- Figure 3 shows data on the 2-, 4- and 8-fold tumour ommended by the university veterinarian department. For regrowth after single and combined treatment with a 10 animals, whose tumours were clamped irradiation was Gy top up dose, exemplarily. In this subset of experi- performed 10 minutes after applying a narrow lace to the ments, five of nine mice were lacking any tumour right hind limb just at the proximal end of the tumour to regrowth 270 days after start of treatment. Analysis of the make the hypoxic radiation conditions as consistent as median time of tumour regrowth after combined treat- possible. Experiments were performed in one run with ment was impaired by an unexpected high rate of local 252 animals. control (figure 3). Therefore, we decided to choose the more complex probit non linear regression analysis. Tumour volumes were scored twice a week, no blinding took place. Follow up was discontinued after 270 days or Figure 4 depicts the extraordinary efficacy of the com- in case of intercurrent death or if tumours had grown to bined treatment by the probit analysis. Irradiation with eight-times the initial tumour volume at the start of treat- graded top up doses from 0 to 44.2 Gy alone resulted in ment. Growth delay and local tumour control were end- local tumour control from 0 to 52% under ambient con- points of the study. All animal experiments were ditions (figure 4a, grey solid line). Addition of lexatumu- accomplished in accordance with the guidelines of the mab after fractionated irradiation alone already caused local authorities (Regional Board Tuebingen, Germany, very high tumour control rates of 85-87%, regardless of appl.no. R4/04) and the German animal welfare regula- the top up dose (p = 0.000006, figure 4a, black solid line). tions. Under clamped bloodflow, treatment with lexatumumab enhanced local tumour control after irradiation with frac- Statistical Analysis tionated irradiation and graded top up doses (0 to 44.2 Statistical analysis was performed as described before[21]. Gy) alone from 0% - 30% (figure 4b, grey solid line) up to In short terms, an exponential regression model was used 43 - 87% (p = 0.00003, figure 4b, black solid line). Statis- to interpolate median tumour regrowth times. Regrowth tical analysis unveiled a highly significant increase of delay was compared by unparametric Kruskal-Wallis tests tumour control rates under both, ambient (p < 0.0001) with Dunn's post tests. Tumour control rates were calcu- and hypoxic (p < 0.0001) conditions (table 1). lated accounting for censored animals as described by Walker and Suit[28]. Data were analysed by a probit non Discussion linear regression analysis. Parameters were estimated Our data prove that the combination of the proapoptotic using the maximum likelihood method. Statistical signif- human antibody lexatumumab with ionizing radiation icance was calculated asymptotically by means of a Hes- has an obvious influence on local tumour control in a sian matrix (STATISTICA 6.0 StatSoft, Hamburg, long-term xenograft model. The effect is evident after irra- Germany). diation with low doses, already. Results It is important to note that these experiments with an ago- Treatment with lexatumumab failed to induce any nistic antibody against TRAIL receptor DR5 corroborate immune reactions of the irradiated skin. No evidence of our recently published data on a high efficacy of a com- acute toxicity was observed. Follow up revealed no signif- bined treatment with another proapoptotic antibody icant differences in frequency of intercurrent deaths after (mapatumumab, anti-DR4) and irradiation. Both models irradiation alone or combined treatment with lexatumu- are in line with in vitro data from our and other labs dem- mab (5.6% vs. 4.6%). onstrating that irradiation acts as a TRAIL sensitizer and not obversely[3,29,30]. Page 3 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 d5 d1 d7 d10 d18 d32 d46 d81 Photograph tu Figure 2 mumab (0.75 mg/kg; ic showcase of the d 1, 4 an chronological d 8) from day 1(d1 sequence of tumour regression and tumou ) up to day 81 (d81) of treatment r regrowth after i.p. application of lexa- Photographic showcase of the chronological sequence of tumour regression and tumour regrowth after i.p. application of lexatumumab (0.75 mg/kg; d 1, 4 and 8) from day 1(d1) up to day 81 (d81) of treatment. Page 4 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 control lexa 10 Gy a 10 Gy a + lexa 0 2040 60270 Follow up [d] Median treatment Figure 3 tumour regrowth times, calculated for two-, four-, and eight-fold tumour size of the initial tumour volume at start of Median tumour regrowth times, calculated for two-, four-, and eight-fold tumour size of the initial tumour vol- ume at start of treatment. Crossbars show 25-75% quartiles for each tumour volume and each treatment. Control; small circle, solid line = animals receiving only i.p. injection with medium, without any further treatment. 10 Gy, square, solid line = fractionated irradiation (3 × 5 Gy) + 10 Gy single top up irradiation. Lexa; triangle, solid line = lexatumumab (0.75 mg/kg body weight, i.p. injection d 1, 4, 8). 10 Gy + lexa; large circle, solid line = fractionated irradiation (3 × 5 Gy) + 10 Gy single top up irradiation and lexatumumab (0.75 mg/kg body weight, i.p. injection d 1, 4, 8). a = Treatment under ambient conditions. This principle diverges from other combined approaches ing a considerable mitochondrial relevance for this syner- where classical chemotherapeutic or other molecular tar- gizing principle[10,39,40]. geted agents act as radiosensitizer. E.g. the synergizing effi- cacy of cisplatin is based on increased oxygenation of The role of radiation induced TRAIL receptor upregulation hypoxic cells and an influence in DNA-repair and cell has been discussed extensively. However, we and others cycle regulation [31-33]. Cetuximab, an antibody against found an only weak or lacking correlation between upreg- epidermal growth factor receptor, seems also to influence ulation and synergism [10,41,42]. Although, other mech- long-term tumour control by affecting DNA damage anisms like cell cycle regulation might play a role [43]. repair[34,35]. It is important to note, that this synergistic principle In contrast to former reports the mitochondrial pathway works under ambient and hypoxic conditions as well. has a strong impact in TRAIL induced apoptosis. Depend- Weinmann et al. demonstrated an undiminished efficacy ing on the cell system applied mitochondrial amplifica- of TRAIL alone under hypoxia in a lymphoma cell tion loops account for its high efficacy[36,37]. In model[44]. Takahashi at al. reported similar observations combination with TRAIL, irradiation increases apoptosis on clonogenic cell kill of A549 cells after treatment with in tumour cells with an impaired mitochondrial pathway. TRAIL and irradiation[45]. However, it remains specula- Furthermore, preirradiation of bcl-2 overexpressing lym- tive why this effect on local tumour control is more pro- phoma cells raises cell death rates after TRAIL receptor nounced under normoxia than under hypoxia. The stimulation[38]. In several tumour cell systems, the proa- known increase of intrinsic radioresistance of hypoxic poptotic molecule Bax was shown to be essential for the cells will be responsible for this reduced susceptibility. combined effect of TRAIL and ionizing radiation suggest- Page 5 of 8 (page number not for citation purposes) relative tumour volume [x-fold tumor size of the initial tumour volume at start of treatment] Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 a b 1 1 0.9 0.9 0.8 0.88 0.7 0.7 0.6 0.6 0.5 0.5 0.4 0.4 0.3 0.3 0.2 0.2 0.1 0.1 0 0 0 10 20 30 40 50 0 10 20 30 40 50 Dose [Gy] Dose [Gy] Dose-r Figure 4 esponse relation between tumour control probability and top up irradiation dose for Colo205 xenograft tumours Dose-response relation between tumour control probability and top up irradiation dose for Colo205 xenograft tumours. Grey circle, solid grey line = tumours treated with fractionated irradiation (5 × 3 Gy) and graded single top up doses (0-44.2 Gy) alone. Black diamond, solid black line = tumours treated with fractionated irradiation (5 × 3 Gy) and graded single top up doses (0-44.2 Gy) and lexatumumab (0.75 mg/kg body weight, i.p. injection d 1, 4, 8) a: under ambient conditions, b: under hypoxic conditions. Dashed lines represent the 95% confidence level. The strong request on the development of personalized an accurate identification of patient collectives who bene- targeted therapies has amazingly changed the general fit from a given treatment. Therefore, a specific subset of approach to cancer treatment. In contrast to cytostatic marker molecules should be identified for each targeted drugs being prescribed on base of classical features as drug [46-48]. TNM classification and histology, targeted drugs require Conclusion Table 1: Results of the probit regression analysis comparing The here presented data provide evidence that the combi- combined treatment (lexatumumab (= lexa, 0.75 mg/kg) and nation of apoptosis inducing antibodies with irradiation irradiation (= RT, 5 × 3 Gy and graded top up doses 0-44.2 Gy) strongly increases long-term tumour control. Since with irradiation alone murine long-term control experiments are the only cur- const. B0 RT-dose (B1) lexa (B2) rently accepted functional approach to simulate the effi- cacy of radiation based treatments the given data are an normoxia optimal scientific base for subsequent clinical trials. Parameter (MLE*) - 1.729 0.028 2.062 Competing interests SE 0.386 0.012 0.343 The authors declare that they have no competing interests. p-value 0.0002 0.0294 <0.0001 Authors' contributions clamped hypoxia PM conceived and drafted the manuscript. DJ and SS car- ried out the animal experiments to the same portion. WB Parameter (MLE) - 2.424 0.035 2.097 SE 0.489 0.013 0.396 performed the statistical analysis. MN participated in the p-value <0.0001 0.0147 <0.0001 statistical analysis and in the drafting of the manuscript. CB contributed to interpretation of the data and critically Regression constant B0 reviewed the article. All authors read and approved the * Maximum likelihood estimate final manuscript. § Standard error Page 6 of 8 (page number not for citation purposes) p Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 tumors or lymphomas.ASCO Annual meeting. Orlando, Acknowledgements Florida, USA, . J Clin Oncol 2008. May 20 suppl; abstr 3537 We thank Human Genome Sciences, Inc. for providing lexatumumab and 15. Sikic BI, Wakelee H, von Mehren M, Lewis NL, Plummer ER, Calvert Dirk Schiller, University of Tübingen, for providing the pictures on tumour AH, Fox NL, Kumm EA, Jones DF, Burris HA: A phase 1b study to growth after treatment with lexatumumab. In addition, we like to thank assess the safety of lexatumumab, a human monoclonal anti- body that activates TRAIL-R2, in combination with gemcit- Katrin Stasch and Stefan Ablasser for technical assistance. This work was abine, pemetrexed, doxorubicin or FOLFIRI. Abstract, 2007. supported by a grant from the Federal Ministry of Education and Research Proceedings of the American Society of Clinical Oncology 25:14006. (Fö: 1456-00) to CB and VJ and by the 'Deutsche Krebshilfe' (Grants10- 16. Tolcher AW, Mita M, Meropol NJ, von Mehren M, Patnaik A, Padavic 1764 Be1 and 10-2220 Be4) to CB, PM and WB. K, Hill M, Mays T, McCoy T, Fox NL, Halpern W, Corey A, Cohen RB: Phase I pharmacokinetic and biologic correlative study of mapatumumab, a fully human monoclonal antibody with References agonist activity to tumor necrosis factor-related apoptosis- 1. Ashkenazi A, Holland P, Eckhardt SG: Ligand-based targeting of inducing ligand receptor-1. J Clin Oncol 2007, 25:1390-1395. apoptosis in cancer: The potential of recombinant human 17. Vulfovich M, Saba N: Mapatumumab, human genome sciences/ apoptosis ligand 2/tumor necrosis factor-related apoptosis- glaxosmithkline/takeda. Curr Opin Mol Ther 2005, 7:502-510. inducing ligand (rhapo2l/trail). J Clin Oncol 2008, 26:3621-3630. 18. Younes A, Vose JM, Zelenetz AD, Smith MR, Burris H, Ansell S, Klein 2. Belka C, Schmid B, Marini P, Durand E, Rudner J, Faltin H, Bamberg J, Kumm E, Czuczman M: Results of a phase 2 trial of HGS-ETR1 M, Schulze-Osthoff K, Budach W: Sensitization of resistant lym- (agonistic human monoclonal antibody to TRAIL receptor phoma cells to irradiation-induced apoptosis by the death 1) in subjects with relapsed/refractory non-hodgkin's lym- ligand trail. Oncogene 2001, 20:2190-2196. phoma (NHL). Blood 2005, 106:489. abstr 3. Marini P, Schmid A, Jendrossek V, Faltin H, Daniel PT, Budach W, 19. Straughn JM Jr, Oliver PG, Zhou T, Wang W, Alvarez RD, Grizzle Belka C: Irradiation specifically sensitises solid tumour cell WE, Buchsbaum DJ: Anti-tumor activity of tra-8 anti-death lines to trail mediated apoptosis. BMC Cancer 2005, 5:5. receptor 5 (DR5) monoclonal antibody in combination with 4. Pan G, O'Rourke K, Chinnaiyan AM, Gentz R, Ebner R, Ni J, Dixit VM: chemotherapy and radiation therapy in a cervical cancer The receptor for the cytotoxic ligand trail. Science 1997, model. Gynecol Oncol 2006, 101:46-54. 276:111-113. 20. Buchsbaum DJ, Zhou T, Grizzle WE, Oliver PG, Hammond CJ, Zhang 5. Walczak H, Miller RE, Ariail K, Gliniak B, Griffith TS, Kubin M, Chin S, Carpenter M, LoBuglio AF: Antitumor efficacy of tra-8 anti- W, Jones J, Woodward A, Le T, Smith C, Smolak P, Goodwin RG, DR5 monoclonal antibody alone or in combination with Rauch CT, Schuh JC, Lynch DH: Tumoricidal activity of tumor chemotherapy and/or radiation therapy in a human breast necrosis factor-related apoptosis-inducing ligand in vivo. Nat cancer model. Clin Cancer Res 2003, 9:3731-3741. Med 1999, 5:157-163. 21. Marini P, Budach W, Niyazi M, Junginger D, Stickl S, Jendrossek V, 6. Camidge DR: An agonist monoclonal antibody directed Belka C: Combination of the pro-apoptotic trail-receptor against death receptor 5/trail-receptor 2 for use in the treat- antibody mapatumumab with ionizing radiation strongly ment of solid tumors. Expert Opin Biol Ther 2008, 8:1167-1176. increases long term tumor control under ambient and 7. Fiveash JB, Gillespie GY, Oliver PG, Zhou T, Belenky ML, Buchsbaum hypoxic conditions. Int J Radiat Oncol Biol Phys 2009, 75:198-202. DJ: Enhancement of glioma radiotherapy and chemotherapy 22. Baumann M, Krause M, Zips D, Eicheler W, Dorfler A, Ahrens J, response with targeted antibody therapy against death Petersen C, Bruchner K, Hilberg F: Selective inhibition of the epi- receptor 5. Int J Radiat Oncol Biol Phys 2008, 71:507-516. dermal growth factor receptor tyrosine kinase by 8. Humphreys R, et al.: HGS-TR2J, a human, agonistic, trail recep- BIBX1382BS and the improvement of growth delay, but not tor-2 monoclonal antibody, induces apoptosis, tumor regres- local control, after fractionated irradiation in human fadu sion and growth inhibition as a single agent in diverse human squamous cell carcinoma in the nude mouse. Int J Radiat Biol solid tumor cell lines. Abstract #204.: 16th EORTC-NCI-AACR Sym- 2003, 79:547-559. posium on Molecular Targets and Cancer Therapeutics. Genevre, Swiss 23. Borst P, Borst J, Smets LA: Does resistance to apoptosis affect clinical response to antitumor drugs? Drug Resist Updat 2001, 9. Ichikawa K, Liu W, Zhao L, Wang Z, Liu D, Ohtsuka T, Zhang H, 4:129-131. Mountz JD, Koopman WJ, Kimberly RP, Zhou T: Tumoricidal 24. Brown JM, Wouters BG: Apoptosis, p53, and tumor cell sensi- activity of a novel anti-human dr5 monoclonal antibody with- tivity to anticancer agents. Cancer Res 1999, 59:1391-1399. out hepatocyte cytotoxicity. Nat Med 2001, 7:954-960. 25. Krause M, Prager J, Zhou X, Yaromina A, Dorfler A, Eicheler W, Bau- 10. Marini P, Denzinger S, Schiller D, Kauder S, Welz S, Humphreys R, mann M: EGFR-TK inhibition before radiotherapy reduces Daniel PT, Jendrossek V, Budach W, Belka C: Combined treat- tumour volume but does not improve local control: Differ- ment of colorectal tumours with agonistic trail receptor ential response of cancer stem cells and nontumourigenic antibodies HGS-ETR1 and HGS-ETR2 and radiotherapy: cells? Radiother Oncol 2007, 83:316-325. Enhanced effects in vitro and dose-dependent growth delay 26. Schmitt CA, Lowe SW: Apoptosis is critical for drug response in vivo. Oncogene 2006, 25:5145-5154. in vivo. Drug Resist Updat 2001, 4:132-134. 11. Mom CH, Sleijfer S, Gietema JA, Fox NL, Piganeau C, Lo L, Uges DRA, 27. Harris AL: Hypoxia-a key regulatory factor in tumour growth. Loos W, de Vries EGE, Verweij J: Mapatumumab, a fully human Nat Rev Cancer 2002, 2:38-47. agonistic monoclonal antibody that targets TRAIL-R1, in 28. Walker AM, Suit HD: Assessment of local tumor control using combination with gemcitabine and cisplatin: A phase 1b censored tumor response data. Int J Radiat Oncol Biol Phys 1983, study in patients with advanced solid malignancies. EORTC- 9:383-386. NCI-AACR Prague, Czech Republic; 2006. 29. Shankar S, Singh TR, Chen X, Thakkar H, Firnin J, Srivastava RK: The 12. Motoki K, Mori E, Matsumoto A, Thomas M, Tomura T, Humphreys sequential treatment with ionizing radiation followed by R, Albert V, Muto M, Yoshida H, Aoki M, Tamada T, Kuroki R, Yosh- trail/apo-2l reduces tumor growth and induces apoptosis of ida H, Ishida I, Ware CF, Kataoka S: Enhanced apoptosis and breast tumor xenografts in nude mice. Int J Oncol 2004, tumor regression induced by a direct agonist antibody to 24:1133-1140. tumor necrosis factor-related apoptosis-inducing ligand 30. Shankar S, Singh TR, Srivastava RK: Ionizing radiation enhances receptor 2. Clin Cancer Res 2005, 11:3126-3135. the therapeutic potential of trail in prostate cancer in vitro 13. Pacey S, Plummer RE, Attard G, Bale C, Calvert AH, Blagden S, Fox and in vivo: Intracellular mechanisms. Prostate 2004, 61:35-49. NL, Corey A, de Bono JS: Phase I and pharmacokinetic study of 31. Douple EB, Richmond RC: Radiosensitization of hypoxic tumor HGS-ETR2, a human monoclonal antibody to TRAIL R2, in cells by cis- and trans-dichlorodiammineplatinum (II). Int J patients with advanced solid malignancies. J Clin Oncol 2005, Radiat Oncol Biol Phys 1979, 5:1369-1372. 23:3055. abstr 32. Hoebers FJ, Pluim D, Verheij M, Balm AJ, Bartelink H, Schellens JH, 14. Saleh MN, Percent I, Wood TE, Posej J, Shah J, Carlisle R, Wojtowicz- Begg AC: Prediction of treatment outcome by cisplatin-DNA Praga S, Forero-Torres A: A phase I study of CS-1008 (human- adduct formation in patients with stage III/IV head and neck ized monoclonal antibody targeting death receptor 5 or squamous cell carcinoma, treated by concurrent cisplatin- DR5), administered weekly to patients with advanced solid radiation (radplat). Int J Cancer 2006, 119:750-756. Page 7 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 33. Chu G: Cellular responses to cisplatin. The roles of DNA- binding proteins and DNA repair. J Biol Chem 1994, 269:787-790. 34. Dittmann K, Mayer C, Rodemann HP: Inhibition of radiation- induced egfr nuclear import by c225 (cetuximab) suppresses DNA-PK activity. Radiother Oncol 2005, 76:157-161. 35. Huang SM, Harari PM: Modulation of radiation response after epidermal growth factor receptor blockade in squamous cell carcinomas: Inhibition of damage repair, cell cycle kinetics, and tumor angiogenesis. Clin Cancer Res 2000, 6:2166-2174. 36. Suliman A, Lam A, Datta R, Srivastava RK: Intracellular mecha- nisms of trail: Apoptosis through mitochondrial-dependent and -independent pathways. Oncogene 2001, 20:2122-2133. 37. Cuello M, Coats AO, Darko I, Ettenberg SA, Gardner GJ, Nau MM, Liu JR, Birrer MJ, Lipkowitz S: N-(4-hydroxyphenyl) retinamide (4HPR) enhances trail-mediated apoptosis through enhancement of a mitochondrial-dependent amplification loop in ovarian cancer cell lines. Cell Death Differ 2004, 11:527-541. 38. Belka C, Schmid B, Marini P, Durand E, Rudner J, Faltin H, Bamberg M, Schulze-Osthoff K, Budach W: Sensitization of resistant lym- phoma cells to irradiation-induced apoptosis by the death ligand TRAIL. Oncogene 2001, 20:2190-2196. 39. von Haefen C, Gillissen B, Hemmati PG, Wendt J, Guner D, Mrozek A, Belka C, Dorken B, Daniel PT: Multidomain Bcl-2 homolog bax but not Bak mediates synergistic induction of apoptosis by TRAIL and 5-FU through the mitochondrial apoptosis pathway. Oncogene 2004, 23:8320-8332. 40. Deng Y, Lin Y, Wu X: TRAIL-induced apoptosis requires Bax- dependent mitochondrial release of smac/diablo. Genes Dev 2002, 16:33-45. 41. Griffith TS, Rauch CT, Smolak PJ, Waugh JY, Boiani N, Lynch DH, Smith CA, Goodwin RG, Kubin MZ: Functional analysis of TRAIL receptors using monoclonal antibodies. J Immunol 1999, 162:2597-2605. 42. Luciano F, Ricci JE, Herrant M, Bertolotto C, Mari B, Cousin JL, Auberger P: T and B leukemic cell lines exhibit different requirements for cell death: Correlation between caspase activation, dff40/dff45 expression, DNA fragmentation and apoptosis in T cell lines but not in Burkitt's lymphoma. Leuke- mia 2002, 16:700-707. 43. Wu F, Hu Y, Long J, Zhou YJ, Zhong YH, Liao ZK, Liu SQ, Zhou FX, Zhou YF, Xie CH: Cytotoxicity and radiosensitization effect of TRA-8 on radioresistant human larynx squamous carcinoma cells. Oncol Rep 2009, 21:461-465. 44. Weinmann M, Marini P, Jendrossek V, Betsch A, Goecke B, Budach W, Belka C: Influence of hypoxia on TRAIL-induced apoptosis in tumor cells. Int J Radiat Oncol Biol Phys 2004, 58:386-396. 45. Takahashi M, Inanami O, Kubota N, Tsujitani M, Yasui H, Ogura A, Kuwabara M: Enhancement of cell death by TNF alpha-related apoptosis-inducing ligand (TRAIL) in human lung carcinoma a549 cells exposed to x rays under hypoxia. J Radiat Res (Tokyo) 2007, 48:461-468. 46. Sturm I, Rau B, Schlag PM, Wust P, Hildebrandt B, Riess H, Haupt- mann S, Dorken B, Daniel PT: Genetic dissection of apoptosis and cell cycle control in response of colorectal cancer treated with preoperative radiochemotherapy. BMC Cancer 2006, 6:124. 47. Mrozek A, Petrowsky H, Sturm I, Kraus J, Hermann S, Hauptmann S, Lorenz M, Dorken B, Daniel PT: Combined p53/Bax mutation results in extremely poor prognosis in gastric carcinoma Publish with Bio Med Central and every with low microsatellite instability. Cell Death Differ 2003, scientist can read your work free of charge 10:461-467. 48. Kallioniemi A: CGH microarrays and cancer. CurrOpin Biotechnol "BioMed Central will be the most significant development for 2008, 19:36-40. disseminating the results of biomedical researc h in our lifetime." Sir Paul Nurse, Cancer Research UK Your research papers will be: available free of charge to the entire biomedical community peer reviewed and published immediately upon acceptance cited in PubMed and archived on PubMed Central yours — you keep the copyright BioMedcentral Submit your manuscript here: http://www.biomedcentral.com/info/publishing_adv.asp Page 8 of 8 (page number not for citation purposes) http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Radiation Oncology Springer Journals

Combined treatment with lexatumumab and irradiation leads to strongly increased long term tumour control under normoxic and hypoxic conditions

Loading next page...
 
/lp/springer-journals/combined-treatment-with-lexatumumab-and-irradiation-leads-to-strongly-PRJChOEiT3

References (52)

Publisher
Springer Journals
Copyright
Copyright © 2009 by Marini et al; licensee BioMed Central Ltd.
Subject
Medicine & Public Health; Oncology; Radiotherapy
eISSN
1748-717X
DOI
10.1186/1748-717X-4-49
pmid
19860913
Publisher site
See Article on Publisher Site

Abstract

Purpose: The combination of ionizing radiation with the pro-apoptotic TRAIL receptor antibody lexatumumab has been shown to exert considerable synergistic apoptotic effects in vitro and in short term growth delay assays. To clarify the relevance of these effects on local tumour control long-term experiments using a colorectal xenograft model were conducted. Materials and methods: Colo205-xenograft bearing NMRI (nu/nu) nude mice were treated with fractionated irradiation (5× 3 Gy, d1-5) and lexatumumab (0.75 mg/kg, d1, 4 and 8). The tumour bearing hind limbs were irradiated with graded single top up doses at d8 under normoxic (ambient) and acute hypoxic (clamped) conditions. Experimental animals were observed for 270 days. Growth delay and local tumour control were end points of the study. Statistical analysis of the experiments included evaluation of tumour regrowth and local tumour control. Results: Combined treatment with irradiation and lexatumumab led to a pronounced tumour regrowth-delay when compared to irradiation alone. The here presented long-term experiments revealed a highly significant rise of local tumour control for normoxic (ambient) (p = 0. 000006) and hypoxic treatment (p = 0. 000030). Conclusion: Our data show that a combination of the pro-apoptotic antibody lexatumumab with irradiation reduces tumour regrowth and leads to a highly increased local tumour control in a nude mouse model. This substantial effect was observed under ambient and more pronounced under hypoxic conditions. related apoptosis inducing ligand) receptor 2 (TRAIL-R2) Background Lexatumumab is a fully human agonistic antibody with a induced apoptosis. Although TRAIL-R2 stimulation alone distinct tumour cell specifity via activation of TRAIL (TNF- is highly effective in a wide range of cancer cell lines, effi- Page 1 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 cacy can be increased by combination with other gyro- Materials and methods static drugs (for review see [1]). We have already shown Animals and tumours Immunodeficient NMRI-(nu/nu)-nude mice were pur- that a combined treatment with TRAIL and irradiation exerts highly synergistic effects regarding apoptosis induc- chased from a specific pathogen free colony at the Univer- tion. This enhanced efficacy was detectable in various sity of Essen (Germany) at the age of 4-6 weeks. Animals solid tumour cell lines and lymphoid tumour cells[2,3]. were kept in an individually ventilated cage rack system (Techniplast, Italy) and fed with sterile high calorie labo- Since discovery of TRAIL and its receptors in 1997 a panel ratory food (Sniff, Germany). Drank water was supple- of agonistic antibodies for TRAIL-receptors R1 and R2 mented by chlorotetracycline and potassium sorbate have been developed and tested in clinical phase I and II acidified to a pH of 3.0 with hydrochloric acid. trials [4-18]. However, up to now only little data are avail- able concerning interaction of agonistic TRAIL receptor The Colo205 tumour cell line (established from a colorec- antibodies and irradiation ([7,19,20]. Besides our recently tal adenocarcinoma) was acquired from ATCC (Bethesda, published report no data on experiments with a combina- MD, USA). In NMRI-(nu/nu)-nude mice Colo205 cells tion of a fully human TRAIL receptor antibody and irradi- form solid, roundly shaped tumours without indication ation have been published[21]. for metastasis. Combining mapatumumab or lexatumumab with irradi- Transplantation and experimental design ation, we have demonstrated that this combination exerts Tumour lumps of about 2 mm diameter from a source strong additive and synergistic effects on apoptosis induc- tumour were implanted subcutaneously into the right tion in vitro and in short-term growth delay experi- hind limb of 6-10 week old animals. Approximately 2-3 ments[10]. However, to proof that induction of apoptosis weeks after transplantation tumour growth was measura- evidently translates into definitive tumour stem cell erad- ble. Tumour size was quantified with calipers in two per- ication long-term experiments with local tumour control pendicular diameters. The tumour volume (V) was as primary endpoint might provide a reliable model for calculated as V = (a × b )/2, where a and b are the long axis clinical potency [22-26]. and the short axis, respectively. Scoring of tumour sizes took place three times per week before start of treatment. Therefore, we decided to perform long-term experiments Body weight was monitored once a week. in a nude mouse xenograft model. As radiation sensitivity becomes affected by limiting intratumoural hypoxia we The median tumour volume at the start of experiments run experiments under both ambient and hypoxic condi- was 116 ± 31 mm . Animals were randomly allocated to tions to mimic realistic tumour conditions[27]. 24 treatment arms (scheme see Figure 1): lexatumumab at day 1, 4 and 8 (0.75 mg/kg body weight intraperitoneally Taken together, our experimental series was designed to (i.p.)) alone, fractioned radiotherapy (5 × 3 Gy within five confirm the striking principle that radiation mediated subsequent days) alone. Single dose top up irradiations TRAIL sensitization effectively increases long-term local (0, 10.0, 14.5, 21.0, 30.4, 44.2 Gy) were performed on day tumour control. 8. Combined treatment was performed at day 1, 4 and 8 with lexatumumab (0.75 mg/kg) (figure 1). Control ani- graded top up dose (0-44,2 Gy) lexatumumab (0.75 mg/kg)/ θθ ±±±± irradiation d1 d4 d5 d8 d270 E Figure 1 xperimental design Experimental design. Small bolt = fractionated irradiation at d 1-5, large bolt = graded top up doses 0-44.2 Gy (under ambi- ent/hypoxic conditions, depending on stratification), small arrowhead: application of lexatumumab (0.75 mg/kg body weight), d = day. Page 2 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 mals were treated only with an i.p. injection of medium Figure 2 shows a chronological sequence of the impressive without antibody or irradiation. tumour regression after treatment with lexatumumab (0.75 mg/kg) for one test animal, exemplarily. Obviously, To minimize toxic side effects and to apply high irradia- tumour growth reduction started after the second applica- tion doses in an easy comparable, time saving schedule we tion i.p., already. However, lacking consolidating irradia- choose a combination of fractionated and graded single tion in this example tumour regrowth is evident four high dose (top up) irradiation. 3 Gy single dose was cho- weeks after start of treatment. sen for fractionated irradiation based on previous experi- ments (Marini et al., Oncogene 2006). Fractionated However, combination of very low doses of irradiation irradiation of tumours was applied in inhalation (Isoflu- with lexatumumab led to an unexpected high local rane) narcosis. Top up irradiation under ambient condi- tumour rate, already. Tumour regrowth after combined tions or under clamped hypoxia was performed with i.p. treatment was observed in less than 50% of the animals. narcosis (fentanyl, midazolam, medetomidine), as rec- Figure 3 shows data on the 2-, 4- and 8-fold tumour ommended by the university veterinarian department. For regrowth after single and combined treatment with a 10 animals, whose tumours were clamped irradiation was Gy top up dose, exemplarily. In this subset of experi- performed 10 minutes after applying a narrow lace to the ments, five of nine mice were lacking any tumour right hind limb just at the proximal end of the tumour to regrowth 270 days after start of treatment. Analysis of the make the hypoxic radiation conditions as consistent as median time of tumour regrowth after combined treat- possible. Experiments were performed in one run with ment was impaired by an unexpected high rate of local 252 animals. control (figure 3). Therefore, we decided to choose the more complex probit non linear regression analysis. Tumour volumes were scored twice a week, no blinding took place. Follow up was discontinued after 270 days or Figure 4 depicts the extraordinary efficacy of the com- in case of intercurrent death or if tumours had grown to bined treatment by the probit analysis. Irradiation with eight-times the initial tumour volume at the start of treat- graded top up doses from 0 to 44.2 Gy alone resulted in ment. Growth delay and local tumour control were end- local tumour control from 0 to 52% under ambient con- points of the study. All animal experiments were ditions (figure 4a, grey solid line). Addition of lexatumu- accomplished in accordance with the guidelines of the mab after fractionated irradiation alone already caused local authorities (Regional Board Tuebingen, Germany, very high tumour control rates of 85-87%, regardless of appl.no. R4/04) and the German animal welfare regula- the top up dose (p = 0.000006, figure 4a, black solid line). tions. Under clamped bloodflow, treatment with lexatumumab enhanced local tumour control after irradiation with frac- Statistical Analysis tionated irradiation and graded top up doses (0 to 44.2 Statistical analysis was performed as described before[21]. Gy) alone from 0% - 30% (figure 4b, grey solid line) up to In short terms, an exponential regression model was used 43 - 87% (p = 0.00003, figure 4b, black solid line). Statis- to interpolate median tumour regrowth times. Regrowth tical analysis unveiled a highly significant increase of delay was compared by unparametric Kruskal-Wallis tests tumour control rates under both, ambient (p < 0.0001) with Dunn's post tests. Tumour control rates were calcu- and hypoxic (p < 0.0001) conditions (table 1). lated accounting for censored animals as described by Walker and Suit[28]. Data were analysed by a probit non Discussion linear regression analysis. Parameters were estimated Our data prove that the combination of the proapoptotic using the maximum likelihood method. Statistical signif- human antibody lexatumumab with ionizing radiation icance was calculated asymptotically by means of a Hes- has an obvious influence on local tumour control in a sian matrix (STATISTICA 6.0 StatSoft, Hamburg, long-term xenograft model. The effect is evident after irra- Germany). diation with low doses, already. Results It is important to note that these experiments with an ago- Treatment with lexatumumab failed to induce any nistic antibody against TRAIL receptor DR5 corroborate immune reactions of the irradiated skin. No evidence of our recently published data on a high efficacy of a com- acute toxicity was observed. Follow up revealed no signif- bined treatment with another proapoptotic antibody icant differences in frequency of intercurrent deaths after (mapatumumab, anti-DR4) and irradiation. Both models irradiation alone or combined treatment with lexatumu- are in line with in vitro data from our and other labs dem- mab (5.6% vs. 4.6%). onstrating that irradiation acts as a TRAIL sensitizer and not obversely[3,29,30]. Page 3 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 d5 d1 d7 d10 d18 d32 d46 d81 Photograph tu Figure 2 mumab (0.75 mg/kg; ic showcase of the d 1, 4 an chronological d 8) from day 1(d1 sequence of tumour regression and tumou ) up to day 81 (d81) of treatment r regrowth after i.p. application of lexa- Photographic showcase of the chronological sequence of tumour regression and tumour regrowth after i.p. application of lexatumumab (0.75 mg/kg; d 1, 4 and 8) from day 1(d1) up to day 81 (d81) of treatment. Page 4 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 control lexa 10 Gy a 10 Gy a + lexa 0 2040 60270 Follow up [d] Median treatment Figure 3 tumour regrowth times, calculated for two-, four-, and eight-fold tumour size of the initial tumour volume at start of Median tumour regrowth times, calculated for two-, four-, and eight-fold tumour size of the initial tumour vol- ume at start of treatment. Crossbars show 25-75% quartiles for each tumour volume and each treatment. Control; small circle, solid line = animals receiving only i.p. injection with medium, without any further treatment. 10 Gy, square, solid line = fractionated irradiation (3 × 5 Gy) + 10 Gy single top up irradiation. Lexa; triangle, solid line = lexatumumab (0.75 mg/kg body weight, i.p. injection d 1, 4, 8). 10 Gy + lexa; large circle, solid line = fractionated irradiation (3 × 5 Gy) + 10 Gy single top up irradiation and lexatumumab (0.75 mg/kg body weight, i.p. injection d 1, 4, 8). a = Treatment under ambient conditions. This principle diverges from other combined approaches ing a considerable mitochondrial relevance for this syner- where classical chemotherapeutic or other molecular tar- gizing principle[10,39,40]. geted agents act as radiosensitizer. E.g. the synergizing effi- cacy of cisplatin is based on increased oxygenation of The role of radiation induced TRAIL receptor upregulation hypoxic cells and an influence in DNA-repair and cell has been discussed extensively. However, we and others cycle regulation [31-33]. Cetuximab, an antibody against found an only weak or lacking correlation between upreg- epidermal growth factor receptor, seems also to influence ulation and synergism [10,41,42]. Although, other mech- long-term tumour control by affecting DNA damage anisms like cell cycle regulation might play a role [43]. repair[34,35]. It is important to note, that this synergistic principle In contrast to former reports the mitochondrial pathway works under ambient and hypoxic conditions as well. has a strong impact in TRAIL induced apoptosis. Depend- Weinmann et al. demonstrated an undiminished efficacy ing on the cell system applied mitochondrial amplifica- of TRAIL alone under hypoxia in a lymphoma cell tion loops account for its high efficacy[36,37]. In model[44]. Takahashi at al. reported similar observations combination with TRAIL, irradiation increases apoptosis on clonogenic cell kill of A549 cells after treatment with in tumour cells with an impaired mitochondrial pathway. TRAIL and irradiation[45]. However, it remains specula- Furthermore, preirradiation of bcl-2 overexpressing lym- tive why this effect on local tumour control is more pro- phoma cells raises cell death rates after TRAIL receptor nounced under normoxia than under hypoxia. The stimulation[38]. In several tumour cell systems, the proa- known increase of intrinsic radioresistance of hypoxic poptotic molecule Bax was shown to be essential for the cells will be responsible for this reduced susceptibility. combined effect of TRAIL and ionizing radiation suggest- Page 5 of 8 (page number not for citation purposes) relative tumour volume [x-fold tumor size of the initial tumour volume at start of treatment] Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 a b 1 1 0.9 0.9 0.8 0.88 0.7 0.7 0.6 0.6 0.5 0.5 0.4 0.4 0.3 0.3 0.2 0.2 0.1 0.1 0 0 0 10 20 30 40 50 0 10 20 30 40 50 Dose [Gy] Dose [Gy] Dose-r Figure 4 esponse relation between tumour control probability and top up irradiation dose for Colo205 xenograft tumours Dose-response relation between tumour control probability and top up irradiation dose for Colo205 xenograft tumours. Grey circle, solid grey line = tumours treated with fractionated irradiation (5 × 3 Gy) and graded single top up doses (0-44.2 Gy) alone. Black diamond, solid black line = tumours treated with fractionated irradiation (5 × 3 Gy) and graded single top up doses (0-44.2 Gy) and lexatumumab (0.75 mg/kg body weight, i.p. injection d 1, 4, 8) a: under ambient conditions, b: under hypoxic conditions. Dashed lines represent the 95% confidence level. The strong request on the development of personalized an accurate identification of patient collectives who bene- targeted therapies has amazingly changed the general fit from a given treatment. Therefore, a specific subset of approach to cancer treatment. In contrast to cytostatic marker molecules should be identified for each targeted drugs being prescribed on base of classical features as drug [46-48]. TNM classification and histology, targeted drugs require Conclusion Table 1: Results of the probit regression analysis comparing The here presented data provide evidence that the combi- combined treatment (lexatumumab (= lexa, 0.75 mg/kg) and nation of apoptosis inducing antibodies with irradiation irradiation (= RT, 5 × 3 Gy and graded top up doses 0-44.2 Gy) strongly increases long-term tumour control. Since with irradiation alone murine long-term control experiments are the only cur- const. B0 RT-dose (B1) lexa (B2) rently accepted functional approach to simulate the effi- cacy of radiation based treatments the given data are an normoxia optimal scientific base for subsequent clinical trials. Parameter (MLE*) - 1.729 0.028 2.062 Competing interests SE 0.386 0.012 0.343 The authors declare that they have no competing interests. p-value 0.0002 0.0294 <0.0001 Authors' contributions clamped hypoxia PM conceived and drafted the manuscript. DJ and SS car- ried out the animal experiments to the same portion. WB Parameter (MLE) - 2.424 0.035 2.097 SE 0.489 0.013 0.396 performed the statistical analysis. MN participated in the p-value <0.0001 0.0147 <0.0001 statistical analysis and in the drafting of the manuscript. CB contributed to interpretation of the data and critically Regression constant B0 reviewed the article. All authors read and approved the * Maximum likelihood estimate final manuscript. § Standard error Page 6 of 8 (page number not for citation purposes) p Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 tumors or lymphomas.ASCO Annual meeting. Orlando, Acknowledgements Florida, USA, . J Clin Oncol 2008. May 20 suppl; abstr 3537 We thank Human Genome Sciences, Inc. for providing lexatumumab and 15. Sikic BI, Wakelee H, von Mehren M, Lewis NL, Plummer ER, Calvert Dirk Schiller, University of Tübingen, for providing the pictures on tumour AH, Fox NL, Kumm EA, Jones DF, Burris HA: A phase 1b study to growth after treatment with lexatumumab. In addition, we like to thank assess the safety of lexatumumab, a human monoclonal anti- body that activates TRAIL-R2, in combination with gemcit- Katrin Stasch and Stefan Ablasser for technical assistance. This work was abine, pemetrexed, doxorubicin or FOLFIRI. Abstract, 2007. supported by a grant from the Federal Ministry of Education and Research Proceedings of the American Society of Clinical Oncology 25:14006. (Fö: 1456-00) to CB and VJ and by the 'Deutsche Krebshilfe' (Grants10- 16. Tolcher AW, Mita M, Meropol NJ, von Mehren M, Patnaik A, Padavic 1764 Be1 and 10-2220 Be4) to CB, PM and WB. K, Hill M, Mays T, McCoy T, Fox NL, Halpern W, Corey A, Cohen RB: Phase I pharmacokinetic and biologic correlative study of mapatumumab, a fully human monoclonal antibody with References agonist activity to tumor necrosis factor-related apoptosis- 1. Ashkenazi A, Holland P, Eckhardt SG: Ligand-based targeting of inducing ligand receptor-1. J Clin Oncol 2007, 25:1390-1395. apoptosis in cancer: The potential of recombinant human 17. Vulfovich M, Saba N: Mapatumumab, human genome sciences/ apoptosis ligand 2/tumor necrosis factor-related apoptosis- glaxosmithkline/takeda. Curr Opin Mol Ther 2005, 7:502-510. inducing ligand (rhapo2l/trail). J Clin Oncol 2008, 26:3621-3630. 18. Younes A, Vose JM, Zelenetz AD, Smith MR, Burris H, Ansell S, Klein 2. Belka C, Schmid B, Marini P, Durand E, Rudner J, Faltin H, Bamberg J, Kumm E, Czuczman M: Results of a phase 2 trial of HGS-ETR1 M, Schulze-Osthoff K, Budach W: Sensitization of resistant lym- (agonistic human monoclonal antibody to TRAIL receptor phoma cells to irradiation-induced apoptosis by the death 1) in subjects with relapsed/refractory non-hodgkin's lym- ligand trail. Oncogene 2001, 20:2190-2196. phoma (NHL). Blood 2005, 106:489. abstr 3. Marini P, Schmid A, Jendrossek V, Faltin H, Daniel PT, Budach W, 19. Straughn JM Jr, Oliver PG, Zhou T, Wang W, Alvarez RD, Grizzle Belka C: Irradiation specifically sensitises solid tumour cell WE, Buchsbaum DJ: Anti-tumor activity of tra-8 anti-death lines to trail mediated apoptosis. BMC Cancer 2005, 5:5. receptor 5 (DR5) monoclonal antibody in combination with 4. Pan G, O'Rourke K, Chinnaiyan AM, Gentz R, Ebner R, Ni J, Dixit VM: chemotherapy and radiation therapy in a cervical cancer The receptor for the cytotoxic ligand trail. Science 1997, model. Gynecol Oncol 2006, 101:46-54. 276:111-113. 20. Buchsbaum DJ, Zhou T, Grizzle WE, Oliver PG, Hammond CJ, Zhang 5. Walczak H, Miller RE, Ariail K, Gliniak B, Griffith TS, Kubin M, Chin S, Carpenter M, LoBuglio AF: Antitumor efficacy of tra-8 anti- W, Jones J, Woodward A, Le T, Smith C, Smolak P, Goodwin RG, DR5 monoclonal antibody alone or in combination with Rauch CT, Schuh JC, Lynch DH: Tumoricidal activity of tumor chemotherapy and/or radiation therapy in a human breast necrosis factor-related apoptosis-inducing ligand in vivo. Nat cancer model. Clin Cancer Res 2003, 9:3731-3741. Med 1999, 5:157-163. 21. Marini P, Budach W, Niyazi M, Junginger D, Stickl S, Jendrossek V, 6. Camidge DR: An agonist monoclonal antibody directed Belka C: Combination of the pro-apoptotic trail-receptor against death receptor 5/trail-receptor 2 for use in the treat- antibody mapatumumab with ionizing radiation strongly ment of solid tumors. Expert Opin Biol Ther 2008, 8:1167-1176. increases long term tumor control under ambient and 7. Fiveash JB, Gillespie GY, Oliver PG, Zhou T, Belenky ML, Buchsbaum hypoxic conditions. Int J Radiat Oncol Biol Phys 2009, 75:198-202. DJ: Enhancement of glioma radiotherapy and chemotherapy 22. Baumann M, Krause M, Zips D, Eicheler W, Dorfler A, Ahrens J, response with targeted antibody therapy against death Petersen C, Bruchner K, Hilberg F: Selective inhibition of the epi- receptor 5. Int J Radiat Oncol Biol Phys 2008, 71:507-516. dermal growth factor receptor tyrosine kinase by 8. Humphreys R, et al.: HGS-TR2J, a human, agonistic, trail recep- BIBX1382BS and the improvement of growth delay, but not tor-2 monoclonal antibody, induces apoptosis, tumor regres- local control, after fractionated irradiation in human fadu sion and growth inhibition as a single agent in diverse human squamous cell carcinoma in the nude mouse. Int J Radiat Biol solid tumor cell lines. Abstract #204.: 16th EORTC-NCI-AACR Sym- 2003, 79:547-559. posium on Molecular Targets and Cancer Therapeutics. Genevre, Swiss 23. Borst P, Borst J, Smets LA: Does resistance to apoptosis affect clinical response to antitumor drugs? Drug Resist Updat 2001, 9. Ichikawa K, Liu W, Zhao L, Wang Z, Liu D, Ohtsuka T, Zhang H, 4:129-131. Mountz JD, Koopman WJ, Kimberly RP, Zhou T: Tumoricidal 24. Brown JM, Wouters BG: Apoptosis, p53, and tumor cell sensi- activity of a novel anti-human dr5 monoclonal antibody with- tivity to anticancer agents. Cancer Res 1999, 59:1391-1399. out hepatocyte cytotoxicity. Nat Med 2001, 7:954-960. 25. Krause M, Prager J, Zhou X, Yaromina A, Dorfler A, Eicheler W, Bau- 10. Marini P, Denzinger S, Schiller D, Kauder S, Welz S, Humphreys R, mann M: EGFR-TK inhibition before radiotherapy reduces Daniel PT, Jendrossek V, Budach W, Belka C: Combined treat- tumour volume but does not improve local control: Differ- ment of colorectal tumours with agonistic trail receptor ential response of cancer stem cells and nontumourigenic antibodies HGS-ETR1 and HGS-ETR2 and radiotherapy: cells? Radiother Oncol 2007, 83:316-325. Enhanced effects in vitro and dose-dependent growth delay 26. Schmitt CA, Lowe SW: Apoptosis is critical for drug response in vivo. Oncogene 2006, 25:5145-5154. in vivo. Drug Resist Updat 2001, 4:132-134. 11. Mom CH, Sleijfer S, Gietema JA, Fox NL, Piganeau C, Lo L, Uges DRA, 27. Harris AL: Hypoxia-a key regulatory factor in tumour growth. Loos W, de Vries EGE, Verweij J: Mapatumumab, a fully human Nat Rev Cancer 2002, 2:38-47. agonistic monoclonal antibody that targets TRAIL-R1, in 28. Walker AM, Suit HD: Assessment of local tumor control using combination with gemcitabine and cisplatin: A phase 1b censored tumor response data. Int J Radiat Oncol Biol Phys 1983, study in patients with advanced solid malignancies. EORTC- 9:383-386. NCI-AACR Prague, Czech Republic; 2006. 29. Shankar S, Singh TR, Chen X, Thakkar H, Firnin J, Srivastava RK: The 12. Motoki K, Mori E, Matsumoto A, Thomas M, Tomura T, Humphreys sequential treatment with ionizing radiation followed by R, Albert V, Muto M, Yoshida H, Aoki M, Tamada T, Kuroki R, Yosh- trail/apo-2l reduces tumor growth and induces apoptosis of ida H, Ishida I, Ware CF, Kataoka S: Enhanced apoptosis and breast tumor xenografts in nude mice. Int J Oncol 2004, tumor regression induced by a direct agonist antibody to 24:1133-1140. tumor necrosis factor-related apoptosis-inducing ligand 30. Shankar S, Singh TR, Srivastava RK: Ionizing radiation enhances receptor 2. Clin Cancer Res 2005, 11:3126-3135. the therapeutic potential of trail in prostate cancer in vitro 13. Pacey S, Plummer RE, Attard G, Bale C, Calvert AH, Blagden S, Fox and in vivo: Intracellular mechanisms. Prostate 2004, 61:35-49. NL, Corey A, de Bono JS: Phase I and pharmacokinetic study of 31. Douple EB, Richmond RC: Radiosensitization of hypoxic tumor HGS-ETR2, a human monoclonal antibody to TRAIL R2, in cells by cis- and trans-dichlorodiammineplatinum (II). Int J patients with advanced solid malignancies. J Clin Oncol 2005, Radiat Oncol Biol Phys 1979, 5:1369-1372. 23:3055. abstr 32. Hoebers FJ, Pluim D, Verheij M, Balm AJ, Bartelink H, Schellens JH, 14. Saleh MN, Percent I, Wood TE, Posej J, Shah J, Carlisle R, Wojtowicz- Begg AC: Prediction of treatment outcome by cisplatin-DNA Praga S, Forero-Torres A: A phase I study of CS-1008 (human- adduct formation in patients with stage III/IV head and neck ized monoclonal antibody targeting death receptor 5 or squamous cell carcinoma, treated by concurrent cisplatin- DR5), administered weekly to patients with advanced solid radiation (radplat). Int J Cancer 2006, 119:750-756. Page 7 of 8 (page number not for citation purposes) Radiation Oncology 2009, 4:49 http://www.ro-journal.com/content/4/1/49 33. Chu G: Cellular responses to cisplatin. The roles of DNA- binding proteins and DNA repair. J Biol Chem 1994, 269:787-790. 34. Dittmann K, Mayer C, Rodemann HP: Inhibition of radiation- induced egfr nuclear import by c225 (cetuximab) suppresses DNA-PK activity. Radiother Oncol 2005, 76:157-161. 35. Huang SM, Harari PM: Modulation of radiation response after epidermal growth factor receptor blockade in squamous cell carcinomas: Inhibition of damage repair, cell cycle kinetics, and tumor angiogenesis. Clin Cancer Res 2000, 6:2166-2174. 36. Suliman A, Lam A, Datta R, Srivastava RK: Intracellular mecha- nisms of trail: Apoptosis through mitochondrial-dependent and -independent pathways. Oncogene 2001, 20:2122-2133. 37. Cuello M, Coats AO, Darko I, Ettenberg SA, Gardner GJ, Nau MM, Liu JR, Birrer MJ, Lipkowitz S: N-(4-hydroxyphenyl) retinamide (4HPR) enhances trail-mediated apoptosis through enhancement of a mitochondrial-dependent amplification loop in ovarian cancer cell lines. Cell Death Differ 2004, 11:527-541. 38. Belka C, Schmid B, Marini P, Durand E, Rudner J, Faltin H, Bamberg M, Schulze-Osthoff K, Budach W: Sensitization of resistant lym- phoma cells to irradiation-induced apoptosis by the death ligand TRAIL. Oncogene 2001, 20:2190-2196. 39. von Haefen C, Gillissen B, Hemmati PG, Wendt J, Guner D, Mrozek A, Belka C, Dorken B, Daniel PT: Multidomain Bcl-2 homolog bax but not Bak mediates synergistic induction of apoptosis by TRAIL and 5-FU through the mitochondrial apoptosis pathway. Oncogene 2004, 23:8320-8332. 40. Deng Y, Lin Y, Wu X: TRAIL-induced apoptosis requires Bax- dependent mitochondrial release of smac/diablo. Genes Dev 2002, 16:33-45. 41. Griffith TS, Rauch CT, Smolak PJ, Waugh JY, Boiani N, Lynch DH, Smith CA, Goodwin RG, Kubin MZ: Functional analysis of TRAIL receptors using monoclonal antibodies. J Immunol 1999, 162:2597-2605. 42. Luciano F, Ricci JE, Herrant M, Bertolotto C, Mari B, Cousin JL, Auberger P: T and B leukemic cell lines exhibit different requirements for cell death: Correlation between caspase activation, dff40/dff45 expression, DNA fragmentation and apoptosis in T cell lines but not in Burkitt's lymphoma. Leuke- mia 2002, 16:700-707. 43. Wu F, Hu Y, Long J, Zhou YJ, Zhong YH, Liao ZK, Liu SQ, Zhou FX, Zhou YF, Xie CH: Cytotoxicity and radiosensitization effect of TRA-8 on radioresistant human larynx squamous carcinoma cells. Oncol Rep 2009, 21:461-465. 44. Weinmann M, Marini P, Jendrossek V, Betsch A, Goecke B, Budach W, Belka C: Influence of hypoxia on TRAIL-induced apoptosis in tumor cells. Int J Radiat Oncol Biol Phys 2004, 58:386-396. 45. Takahashi M, Inanami O, Kubota N, Tsujitani M, Yasui H, Ogura A, Kuwabara M: Enhancement of cell death by TNF alpha-related apoptosis-inducing ligand (TRAIL) in human lung carcinoma a549 cells exposed to x rays under hypoxia. J Radiat Res (Tokyo) 2007, 48:461-468. 46. Sturm I, Rau B, Schlag PM, Wust P, Hildebrandt B, Riess H, Haupt- mann S, Dorken B, Daniel PT: Genetic dissection of apoptosis and cell cycle control in response of colorectal cancer treated with preoperative radiochemotherapy. BMC Cancer 2006, 6:124. 47. Mrozek A, Petrowsky H, Sturm I, Kraus J, Hermann S, Hauptmann S, Lorenz M, Dorken B, Daniel PT: Combined p53/Bax mutation results in extremely poor prognosis in gastric carcinoma Publish with Bio Med Central and every with low microsatellite instability. Cell Death Differ 2003, scientist can read your work free of charge 10:461-467. 48. Kallioniemi A: CGH microarrays and cancer. CurrOpin Biotechnol "BioMed Central will be the most significant development for 2008, 19:36-40. disseminating the results of biomedical researc h in our lifetime." Sir Paul Nurse, Cancer Research UK Your research papers will be: available free of charge to the entire biomedical community peer reviewed and published immediately upon acceptance cited in PubMed and archived on PubMed Central yours — you keep the copyright BioMedcentral Submit your manuscript here: http://www.biomedcentral.com/info/publishing_adv.asp Page 8 of 8 (page number not for citation purposes)

Journal

Radiation OncologySpringer Journals

Published: Oct 27, 2009

There are no references for this article.